WO2019096397A1 - Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder - Google Patents

Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder Download PDF

Info

Publication number
WO2019096397A1
WO2019096397A1 PCT/EP2017/079506 EP2017079506W WO2019096397A1 WO 2019096397 A1 WO2019096397 A1 WO 2019096397A1 EP 2017079506 W EP2017079506 W EP 2017079506W WO 2019096397 A1 WO2019096397 A1 WO 2019096397A1
Authority
WO
WIPO (PCT)
Prior art keywords
salt
compound
medicament
administration
dose
Prior art date
Application number
PCT/EP2017/079506
Other languages
English (en)
French (fr)
Inventor
Udai BANERJI
Original Assignee
The Institute Of Cancer Research: Royal Cancer Hospital
The Royal Marsden Nhs Foundation Trust
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Institute Of Cancer Research: Royal Cancer Hospital, The Royal Marsden Nhs Foundation Trust filed Critical The Institute Of Cancer Research: Royal Cancer Hospital
Priority to RU2020119399A priority Critical patent/RU2760006C1/ru
Priority to PCT/EP2017/079506 priority patent/WO2019096397A1/en
Priority to PT187248927T priority patent/PT3709996T/pt
Priority to CN201880073974.9A priority patent/CN111542317A/zh
Priority to KR1020207017064A priority patent/KR102669822B1/ko
Priority to AU2018369325A priority patent/AU2018369325A1/en
Priority to CA3082619A priority patent/CA3082619A1/en
Priority to SG11202004376TA priority patent/SG11202004376TA/en
Priority to MX2020005006A priority patent/MX2020005006A/es
Priority to BR112020009452-8A priority patent/BR112020009452A2/pt
Priority to EP18724892.7A priority patent/EP3709996B1/en
Priority to DK18724892.7T priority patent/DK3709996T3/da
Priority to EP24159280.7A priority patent/EP4364807A2/en
Priority to US16/764,242 priority patent/US11400090B2/en
Priority to PCT/EP2018/062805 priority patent/WO2019096449A1/en
Priority to FIEP18724892.7T priority patent/FI3709996T3/fi
Priority to TW107116587A priority patent/TWI795405B/zh
Publication of WO2019096397A1 publication Critical patent/WO2019096397A1/en
Priority to US17/850,463 priority patent/US20230012116A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • A61K31/37Coumarins, e.g. psoralen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to a therapeutic or prophylactic medicament for a cell proliferative disorder, particularly cancer, comprising a coumarm derivative.
  • a potassium salt of compound (I) is known to be administered to patients with solid cancers such as non-small-cell lung cancer, ovarian cancer, endometrial cancer, and colorectal cancer twice weekly at a dose of 4 mg per administration (see non patent document 1),
  • Patent document 1 WO 2007/091736
  • Patent document 2 WO 2009/014100
  • Non patent document 1 Journal of Clinical Oncology 34, no. 15 restrictive suppl (May 2016) 2582-2582
  • the present invention has been made in light of such circumstances. It is an object of the present invention to provide a dosing regimen for compound (I) or a salt thereof that can be implemented safely and for long periods, as well as a therapeutic or prophylactic medicament for a cell proliferative disorder (particularly cancer) that is used based on such a dosing regimen.
  • the present invention provides medicaments according to the following items A1 to A15.
  • a medicament for the treatment or prevention of a cell proliferative disorder comprising as an active ingredient a compound represented by formula (I):
  • steps (a) and (b) are subsequently repeated at least once.
  • A2 The medicament according to item Al, comprising a potassium salt of a compound represented by formula (I) as an active ingredient.
  • A3 The medicament according to item A1 or A2, wherein the cell proliferative disorder is cancer.
  • A4 The medicament according to any one of items Al to A3, wherein the cell proliferative disorder is a KRAS mutant cancer.
  • A5 The medicament according to any one of items Al to A4, wherein the cell proliferative disorder is a solid cancer.
  • A6 The medicament according to any one of items Al to ⁇ 5, wherein the dose per administration in step (a) is 3.2 mg.
  • A7 The medicament according to item A6, wherein the medicament is used in such a manner that prior to step (a):
  • said compound or salt is administered twice weekly at a dose of 3.2 mg per administration;
  • steps (2a) and (2b) are subsequently repeated at least once.
  • A8 The medicament according to item A7, wherein the medicament is used in such a manner that prior to step (1) or (2), said compound or salt is administered twice weekly at a dose of 4 mg per administration.
  • A9 The medicament according to any one of items A1 to A5 :
  • steps (B2a) and (B2b) are subsequently repeated at least once;
  • A10 The medicament according to any one of items A1 to A4, wherein the cell proliferative disorder is multiple myeloma.
  • Al l The medicament according to item A 10, wherein the dose per administration in step (a) is 4 mg.
  • A12 The medicament according to item A10 or All, wherein the cell proliferative disorder is an NRAS mutant cancer.
  • A13 The medicament according to any one of items A10 to A12, wherein the medicament is used in combination with dexamethasone, and wherein the medicament is used in such a maoner that said compound or salt is administered before, simultaneously with or after administration of dexamethasone.
  • A14 The medicament according to item A13, wherein dexamethasone is administered once weekly at a dose of 20 mg per administration.
  • A15 The medicament according to any one of items A1 to A14, wherein the administration of said compound or salt is oral administration.
  • the medicament of the present invention may consist of compound (I) or a salt thereof, or it may be a pharmaceutical composition further comprising another component.
  • medicaments according to the following items A16 and A17.
  • A16 A medicament for the treatment or prevention of a cell proliferative disorder, the medicament comprising a compound represented by formula (I):
  • the medicament is packaged together with: (i) a container for housing said medicament, and
  • steps (a) and (b) are subsequently repeated at least once.
  • a 17 The medicament according to item A16, wherein the medicament is a medicament according to any one of A1 to A15, and wherein the instruction is an instruction for using said medicament in such a manner that said compound or salt is administered according to the prescribed dosing regimen corresponding to the medicament used.
  • Bl A method for the treatment or prevention of a cell proliferative disorder, the method comprising:
  • B2 The method according to item B 1 , wherein a potassium salt of a compound represented by formula (I) is administered.
  • B3 The method according to item B1 or B2, wherein the cell proliferative disorder is cancer.
  • B5 The method according to any one of items B 1 to B4, wherein the cell proliferative disorder is a solid cancer.
  • step (a) The method according to any one of items B1 to B5, wherein the dose per administration in step (a) is 3.2 mg.
  • B8 The method according to item B7, wherein the method comprises, prior to step (1) or (2), administering said compound or salt twice weekly at a dose of 4 mg per administration.
  • Bll The method according to item BIO, wherein the dose per administration in step (a) is 4 mg.
  • B12 The method according to item BIO or Bll, wherein the cell proliferative disorder is an NRAS mutant cancer.
  • B13 The method according to any one of items BIO to B12, wherein said compound or salt is used in combination with dexamethasone, and wherein the method comprises administering said compound or salt before, simultaneously with or after administration of dexamethasone.
  • B14 The method according to item B13, wherein dexamethasone is administered once weekly at a dose of 20 mg per administration.
  • B15 The method according to any one of items B1 to B14, wherein the administration of said compound or salt is oral administration.
  • steps (a) and (b) are subsequently repeated at least once.
  • C3 The use according to item Cl or C2, wherein the cell proliferative disorder is cancer.
  • said compound or salt is administered twice weekly at a dose of 3.2 mg per administration; or
  • steps (2a) and (2b) are subsequently repeated at least once.
  • C8 The use according to item Cl, wherein the medicament is used in such a maimer that prior to step (1) or (2), said compound or salt is administered twice weekly at a dose of 4 mg per administration.
  • steps (B2a) and (B2b) are subsequently repeated at least once;
  • step (a) The use according to item C10, wherein the dose per administration in step (a) is 4 mg. Cl 2; The use according to item C10 or Cll, wherein the cell proliferative disorder is an NRAS mutant cancer,
  • the dosing regimen that is used in the present invention comprises repeating a cycle comprising a prescribed rest period, and makes it possible to administer compound (1) or a salt thereof for long periods while minimizing side effects and maintaining the drug efficacy.
  • the dosing regimen makes it possible to treat or prevent cell proliferative disorders, particularly cancer, while minimizing the burden on patients.
  • a dosing regimen for compound (I) or a salt thereof that can be implemented safely and for long periods, as well as a therapeutic or prophylactic medicament for a cell proliferative disorder (particularly cancer) that is used based on such a dosing regimen.
  • Fig. 1 is a graph showing change in tumor size over time in a KRAS mutant non-small-cell lung cancer patient who received treatment by the medicament of the present invention.
  • the horizontal axis (X-axis) represents the treatment cycle number and the vertical axis (Y-axis) represents the rate of change of tumor size from baseline.
  • the tumor size was measured by CT scan.
  • Compound (I) and salts thereof can be prepared by the method described in WO 2007/091736 or WO 2013/035754.
  • the active ingredient to be used in the present invention is preferably a pharmaceutically acceptable salt of compound (I).
  • salts include: inorganic acid salts such as hydrochlorides, hydrobromides, hydroiodides, sulfates and phosphates; sulfonates such as methanesulfonates, benzenesulfonates and toluenesulfonates; carboxylates such as formates, acetates, oxalates, maleates, fumarates, citrates, malates, succinates, malonates, gluconates, mandelates, benzoates, salicylates, fiuoroacetates, trifiuoroacetates, tartrates, propionates and glutarates; alkali metal salts such as lithium salts, sodium salts, potassium salts, cesium salts and rubidium salts; alkaline earth metal salts such as magnesium salts and calcium salts; and ammonium salts
  • alkali metal salts such as lithium salts, sodium salts, potassium salts, cesium salts and rubidium salts are preferred, sodium salts and potassium salts are more preferred, and potassium salts are particularly preferred.
  • Specific examples of potassium salts of compound (I) include a salt represented by the following formula (la).
  • Examples of cell proliferative disorders to be treated or prevented by the medicament or method of the present invention include cancer, rheumatism and inflammation, among which cancer is preferred.
  • cancers include: blood and lymphoid cancers, such as leukemias (acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia, and the like), malignant lymphomas (Hodgkin’s disease, non-Hodgkin’s lymphoma, and the like), multiple myeloma, and myelodysplastic syndrome; central nervous system cancers, such as brain tumor and glioma; and solid cancers, such as head and neck cancers (pharyngeal cancer, laryngeal cancer, tongue cancer, and the like), esophageal cancer, gastric cancer, colorectal cancer (cecal cancer, colon cancer, rectal cancer, or the like), lung cancer (small cell lung cancer, non-small cell lung cancer, or the like), thyroid cancer, breast cancer, gallbladder cancer, pancreatic cancer, liver cancer, prostate cancer, ovarian cancer, uterine cancer (
  • a preferred blood or lymphoid cancer is multiple myeloma.
  • preferred solid cancers include ovarian cancer, breast cancer, uterine cancer, colorectal cancer, and lung cancer, among which non-small cell lung cancer is particularly preferred.
  • Preferred cancers are multiple myeloma and solid cancers, among which multiple myeloma and non-small cell lung cancer are particularly preferred.
  • the cancer may be one with a gene mutation or without a gene mutation, or one where the presence or absence of mutation is unclear, but it is preferably one with a gene mutation.
  • genes to be mutated include EGFR, FGFR, ALK, ROS1, PI3K, BRAF, KRAS and NRAS.
  • the cancer is preferably a KRAS mutant and/or NRAS mutant one, and more preferably it is KRAS mutant and NRAS mutant multiple myeloma or a KRAS mutant solid cancer (particularly non-small-cell lung cancer).
  • the subject to be administered compound (I) or a salt thereof is an animal, preferably a mammal (for example, a mouse, a rat, a rabbit, a dog, a monkey (for example, a cynomolgus monkey), or a human), and most preferably a human.
  • the human may be an adult (18 years or older) or a child (younger than 18). In the case of a child, it is preferably one of age at least 6 months or older, for example.
  • systemic administration such as oral administration, rectal administration, intravenous administration, intramuscular administration, subcutaneous administration, intracistemal administration, vaginal administration, intraperitoneal administration, intravesical administration or inhalation administration; or topical administration in the form of an ointment, gel, cream or the like. Oral administration is preferred.
  • Compound (I) or a salt thereof is generally prepared as a certain formulation (dosage form).
  • the formulation may be, for example, a tablet, a capsule, a granule, a powder, a fine granule, a pill, or an aqueous or nonaqueous solution or suspension.
  • the solution or suspension may be stored filled in a container suited for the preparation of an individual dose.
  • Each of such formulations as the ones mentioned above may be produced by a known method, by mixing the compound (I) or salt thereof with a pharmaceutically acceptable additive.
  • additives include excipients, lubricants (coating agents), binders, disintegrants, stabilizers, flavoring agents, bases, dispersants, diluents, surfactants, emulsifiers, and the like.
  • excipients examples include starches (starch, potato starch, maize starch and the like), lactose, crystalline cellulose, and calcium hydrogen phosphate.
  • lubricants examples include ethyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, shellac, talc, camauba wax, and paraffin.
  • binders examples include polyvinylpyrrolidone and macrogol, as well as the same compounds as mentioned for the excipient.
  • disintegrants include chemically modified starches and celluloses, such as croscarmellose sodium, sodium carboxymethyl starch, and crosslinked polyvinylpyrrolidone, as well as the same compounds as mentioned for the excipient.
  • stabilizers include: paraoxybenzoic acid esters such as methylparaben and propylparaben; benzalkonium chloride; phenols such as phenol and cresol; thimerosal; dehydroacetic acid; and sorbic acid.
  • flavoring agents include sweeteners, acidulants and fragrances which are commonly used.
  • bases include: fats such as lard; vegetable oils such as olive oil and sesame oil; higher alcohols such as stearyl alcohol and cetanol; animal oils; lanolin acid; vaseline; paraffins; bentonite; glycerine; and glycol oils.
  • dispersants include cellulose derivatives (gum arabic, tragacanth, methyl cellulose and the like), stearic add polyesters, sorbitan sesquioleate, aluminum monostearate, sodium alginate, polysorbates, and sorbitan fatty acid esters,
  • solvents and diluents in liquid formulations include phenol, chlorocresol, purified water and distilled water.
  • surfactants and emulsifiers examples include polysorbate 80, polyoxyl 40 stearate, and lauromacrogol.
  • the preferred percentage of the compound (I) or salt thereof contained in the formulation will differ depending on the dosage form, but it is generally 0.01% to 100% by weight with respect to the total weight of the formulation.
  • the content of the compound (I) or salt thereof in the formulation may be set as appropriate for the predetermined dosage.
  • the preferred content is 0.01 mg to 10 mg, for example; for a capsule, it may be 0.1 mg to 4 mg, for example.
  • a more preferred content is 0.8 mg, for example.
  • compound (I) or a salt thereof is administered in the following manner:
  • steps (a) and (b) are subsequently repeated at least once.
  • to be administered“twice weekly 3 ’ means that the compound (I) or salt thereof is administered two times during a one-week period. Administration may be performed twice on the same day, or once a day on different days (which may be consecutive), but it is preferably performed on different days. More preferably, administration is performed on the 1st and 4th days or the 3rd and 6th days of the period, for example, so that the compound (I) or salt thereof is administered at dose intervals as uniform as possible, i.e., at dose intervals of 3 to 4 days.
  • the one-week period may start on a Monday, for example, or it may start on a Wednesday, for example.
  • When two administrations are performed on different days each may be performed at any time of day, but they are preferably performed at the same time of day (for example, after breakfast).
  • the dose of the compound (I) or salt thereof per administration is preferably 3.2 mg or 4 mg.
  • the dose is preferably 4 mg, and when the cancer is a solid cancer (particularly non-small-cell lung cancer), it is preferably 3.2 mg or 4 mg,
  • the 4-week cycle consisting of steps (a) and (b) is repeated two times (8 weeks) to 60 times (approximately 4 years and 8 months), for example, and more specifically, it is repeated 8 times (32 weeks) or 18 times (72 weeks), for example. Even if the number of cycles to be repeated has previously been determined, the number of cycles may be changed based on the judgment of the physician or veterinarian depending on, for example, the condition of the subject. Moreover, administration may even be stopped during the cycle based on the judgment of the physician or veterinarian depending on, for example, the condition of the subject.
  • the compound (I) or salt thereof is administered twice weekly at a dose of 3.2 mg per administration;
  • steps (2a) and (2b) are subsequently repeated at least once.
  • step (1) or (2) Moreover, optionally, prior to step (1) or (2),
  • the cell proliferative disorder to be treated or prevented based on the dosing regimen of this embodiment is preferably a solid cancer and more preferably non-small-cell lung cancer.
  • a cycle consisting of 4 weeks of administration is usually repeated two times (8 weeks) to 30 times (approximately 2 years and 4 months), and it is preferably repeated 13 times (52 weeks), for example.
  • step (3) a cycle consisting of 4 weeks of administration is usually repeated two times (8 weeks) to 30 times (approximately 2 years and 4 months), and it is preferably repeated 8 times (32 weeks), for example.
  • steps (1) and (3) even if the number of cycles to be repeated has previously been determined, the number of cycles may be changed depending on the judgment of the physician or veterinarian depending on, for example, the condition of the subject. Moreover, administration may even be stopped during the cycle based on the judgment of the physician or veterinarian depending on, for example, the condition of the subject.
  • compound (I) or a salt thereof is administered in the following manner:
  • steps (B2a) and (B2b) are subsequently repeated at least once;
  • the cell proliferative disorder to be treated or prevented based on the dosing regimen of this embodiment is preferably cancer and more preferably a solid cancer.
  • solid cancers non-small cell lung cancer is preferred, and KRAS mutant non-small cell lung cancer is particularly preferred.
  • step (A) a cycle consisting of 4 weeks of administration is usually repeated two times (8 weeks) to 30 times (approximately 2 years and 4 months), and it is preferably repeated 13 times (52 weeks), for example.
  • step (Bl) a cycle consisting of 4 weeks of administration is usually repeated two times (8 weeks) to 30 times (approximately 2 years and 4 months), and it is preferably repeated 8 times (32 weeks), for example.
  • steps (A), (Bl), (B2) and ( €) even if the number of cycles to be repeated has previously been determined, the number of cycles may be changed depending on the judgment of the physician or veterinarian depending on, for example, the condition of the subject. Moreover, administration may even be stopped during the cycle based on the judgment of the physician or veterinarian depending on, for example, the cond ition of the subject.
  • Compound (I) or a salt thereof may be used either alone or in combination with another drug.
  • the other drug may be an antiemetic drug or anticancer agent, for example, and is preferably an anticancer agent.
  • anticancer agents include thalidomide-based anticancer agents such as thalidomide and lenalidomide, proteasome inhibitors such as bortezomib and ixazomib, and steroidal anticancer agents such as dexamethasone and prednisolone.
  • cancer is non-small-cell lung cancer
  • compound (I) or a salt thereof is preferably used alone.
  • the other drag is preferably dexamethasone, for example.
  • Dexamethasone is a drag that is part of the standard of care for multiple myeloma.
  • dexamethasone is preferably administered once weekly and the dose of dexamethasone per administration is preferably, for example, 20 mg.
  • compound (I) or a salt thereof is used in combination with dexamethasone
  • the order and timing of their administrations are not particularly restricted, and the compound (I) or salt thereof may be administered before, simultaneously with or after administration of dexamethasone.
  • step (a) when the compound (I) or salt thereof and dexamethasone are administered in a one-week period, it is preferred, for example, that; the compound (!) or salt thereof is administered on the 1st and 4th days and dexamethasone on the 2nd day of the period, or the compound (I) or salt thereof is administered on the 3rd and 6th days and dexamethasone on the 4th day of the period, or the compound (I) or salt thereof is administered on the 2nd and 5th days and dexamethasone on the 1st day of the period, or the compound (I) or salt thereof is administered on the 4th and 7th days and dexamethasone on the 2nd day of the period.
  • the one-week period may start on a Monday, for example, or it may start on a Wednesday, for example, Each administration may be performed at any time of day, but the administrations are preferably performed at the same time of day (for example, after dinner).
  • the period during which the compound (I) or salt thereof is used in combination with dexamethasone may be determined based on the judgment of the physician or veterinarian depending on, for example, die condition of the subject * Also, administration of either or both the compound (I) or salt thereof and dexamethasone may be stopped based on the judgment of the physician or veterinarian depending on, for example, the condition of the subject.
  • a medicament for the treatment or prevention of a cell proliferative disorder that comprises a compound represented by formula (I):
  • steps (a) and (b) are subsequently repeated at least once.
  • the packaged medicament is a medicament according to any one of items A1 to A15 above and that the instruction ⁇ ) above is/are an instmction(s) for using the medicament in such a maimer that the compound (I) or salt thereof is administered according to the prescribed dosing regimen corresponding to the medicament used.
  • the phrase “prescribed dosing regimen corresponding to the medicament used” means, in the case of the medicament of item A9, for example, any of the dosing regimens (Rl) to (R3) prescribed in item A9.
  • the packaging may be performed using a packaging material (such as a carton), for example.
  • the package may also include a label, a pamphlet or a pharmaceutically acceptable cushioning material , for example.
  • the container of (i) above is, for example, a bottle or a FTP sheet, and it may be produced from a material such as glass, plastic or aluminum, for example.
  • the container is printed with leters indicating, for example, that the medicament is to be used for treatment or prevention of the prescribed disease, or it has a label atached on which such letters are printed.
  • the medicament may or may not be housed in the container of (i), but is preferably housed in it.
  • the instmction(s) of (it) above include(s) information necessary for use of the medicament based on the prescribed dosing regimen.
  • the instruction(s) may also include information relating to the efficacy and effects of the medicament, for example.
  • the instructions) of (ii) may be in die form of a document, for example.
  • the document containing the mstruction(s) may be a package insert, for example.
  • the document containing the instruction(s) may also be printed on a label, or printed on a packaging material (such as a carton), for example.
  • the mstruction(s) may also be in the form of printing on paper or plastic, for example, or in the form of electronic storage in a storage medium such as a CD-ROM or flash memory.
  • a potassium salt of compound (I) (the potassium salt being also referred to as the“IMP” in this example) was administered to a patient with KRAS mutant and NRAS mutant IgG lambda multiple myeloma at a dose of 4 mg twice weekly (Tuesday and Friday) in 4-week cycles consisting of 3 weeks of treatment followed by 1 week of rest (3 weeks on/ 1 week off). Dexamethasone was not used in combination with the IMP.
  • a potassium salt of compound (I) (the potassium salt being also referred to as the“IMP” in this example) was administered to a patient with KRAS mutant non-small-cell lung cancer.
  • the IMP was first administered at a dose of 4 mg twice weekly in 4-week cycles consisting of 4 weeks of treatment (4mg 4wk on).
  • Cycle 9 due to Grade 3 maculopapular rash, the dosage was reduced to 3.2 mg twice weekly in 4- week cycles (3.2mg 4wk on).
  • Cycle 22 due to Grade 3 facial rash, the dosage was reduced again to a 3 weeks on/ 1 week off schedule with 3.2 mg twice weekly (3.2mg 3wk on/ Iwk off).
  • PR partial response
  • RECIST Response Evaluation Criteria in Solid Tumors
  • Fig. 1 The change in tumor size in the patient over the first 40 cycles (approximately 3 years) is shown in Fig. 1.
  • the horizontal axis (X-axis) represents the treatment cycle number and the vertical axis
  • Y-axis represents the rate of change of tumor size from baseline.
  • the tumor size was measured by CT scan.
  • the best responses of the dosing schedules 4mg 4wk on, 3,2mg 4wk on, and 3.2mg 3wk on/ Iwk off were 58% reduction, 61% reduction, and 68% reduction in target lesions by RECIST 1.1, respectively.
  • Treatment of the patient by the IMP is being continued as of October 2017 (at Cycle 42)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Oncology (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
PCT/EP2017/079506 2017-11-16 2017-11-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder WO2019096397A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
RU2020119399A RU2760006C1 (ru) 2017-11-16 2017-11-16 Производное кумарина для лечения или профилактики расстройства пролиферации клеток
PCT/EP2017/079506 WO2019096397A1 (en) 2017-11-16 2017-11-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder
BR112020009452-8A BR112020009452A2 (pt) 2017-11-16 2018-05-16 Derivado de cumarina para terapia ou profilaxia de um distúrbio proliferativo celular
DK18724892.7T DK3709996T3 (da) 2017-11-16 2018-05-16 Coumarin-derivat til behandling eller forebyggelse af en celleproliferativ lidelse
KR1020207017064A KR102669822B1 (ko) 2017-11-16 2018-05-16 세포 증식성 질환의 치료 또는 예방을 위한 쿠마린 유도체
AU2018369325A AU2018369325A1 (en) 2017-11-16 2018-05-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder
CA3082619A CA3082619A1 (en) 2017-11-16 2018-05-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder
SG11202004376TA SG11202004376TA (en) 2017-11-16 2018-05-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder
MX2020005006A MX2020005006A (es) 2017-11-16 2018-05-16 Derivado de cumarina para terapia o profilaxis de un trastorno proliferativo celular.
PT187248927T PT3709996T (pt) 2017-11-16 2018-05-16 Derivado de coumarina para terapia ou profilaxia de um distúrbio de proliferação celular
EP18724892.7A EP3709996B1 (en) 2017-11-16 2018-05-16 Coumarin derivative for therapy or prophylaxis of a disorder of cell proliferation
CN201880073974.9A CN111542317A (zh) 2017-11-16 2018-05-16 用于治疗或预防细胞增殖性疾病的香豆素衍生物
EP24159280.7A EP4364807A2 (en) 2017-11-16 2018-05-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder
US16/764,242 US11400090B2 (en) 2017-11-16 2018-05-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder
PCT/EP2018/062805 WO2019096449A1 (en) 2017-11-16 2018-05-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder
FIEP18724892.7T FI3709996T3 (fi) 2017-11-16 2018-05-16 Kumariinijohdannainen soluproliferaation häiriön hoitamiseksi tai ennaltaehkäisemiseksi
TW107116587A TWI795405B (zh) 2017-11-16 2018-05-16 香豆素衍生物用於製備治療或預防細胞增殖性病症之藥物之用途
US17/850,463 US20230012116A1 (en) 2017-11-16 2022-06-27 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2017/079506 WO2019096397A1 (en) 2017-11-16 2017-11-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder

Publications (1)

Publication Number Publication Date
WO2019096397A1 true WO2019096397A1 (en) 2019-05-23

Family

ID=60331633

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2017/079506 WO2019096397A1 (en) 2017-11-16 2017-11-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder
PCT/EP2018/062805 WO2019096449A1 (en) 2017-11-16 2018-05-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/062805 WO2019096449A1 (en) 2017-11-16 2018-05-16 Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder

Country Status (14)

Country Link
US (2) US11400090B2 (zh)
EP (2) EP4364807A2 (zh)
CN (1) CN111542317A (zh)
AU (1) AU2018369325A1 (zh)
BR (1) BR112020009452A2 (zh)
CA (1) CA3082619A1 (zh)
DK (1) DK3709996T3 (zh)
FI (1) FI3709996T3 (zh)
MX (1) MX2020005006A (zh)
PT (1) PT3709996T (zh)
RU (1) RU2760006C1 (zh)
SG (1) SG11202004376TA (zh)
TW (1) TWI795405B (zh)
WO (2) WO2019096397A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11517573B2 (en) 2019-09-13 2022-12-06 The Institute Of Cancer Research: Royal Cancer Hospital Therapeutic compositions, combinations, and methods of use
US11873296B2 (en) 2022-06-07 2024-01-16 Verastem, Inc. Solid forms of a dual RAF/MEK inhibitor

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007091736A1 (ja) 2006-02-09 2007-08-16 Chugai Seiyaku Kabushiki Kaisha 抗腫瘍活性を有する新規クマリン誘導体
WO2009014100A1 (ja) 2007-07-20 2009-01-29 Chugai Seiyaku Kabushiki Kaisha p27蛋白質誘導剤
WO2013035754A1 (ja) 2011-09-05 2013-03-14 中外製薬株式会社 クマリン誘導体の製造方法

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9920908D0 (en) 1999-09-03 1999-11-10 Indena Spa Chalcone coumarins

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007091736A1 (ja) 2006-02-09 2007-08-16 Chugai Seiyaku Kabushiki Kaisha 抗腫瘍活性を有する新規クマリン誘導体
US20110092700A1 (en) * 2006-02-09 2011-04-21 Iikura Hitoshi Novel Coumarin Derivative Having Antitumor Activity
WO2009014100A1 (ja) 2007-07-20 2009-01-29 Chugai Seiyaku Kabushiki Kaisha p27蛋白質誘導剤
WO2013035754A1 (ja) 2011-09-05 2013-03-14 中外製薬株式会社 クマリン誘導体の製造方法

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHENARD-POIRIER ET AL: "Meeting Library | Results from the biomarker-driven basket trial of RO5126766 (CH5127566), a potent RAF/MEK inhibitor, in RAS- or RAF-mutated malignancies including multiple myeloma.", 3 June 2017 (2017-06-03), XP055495510, Retrieved from the Internet <URL:https://meetinglibrary.asco.org/record/144582/abstract> [retrieved on 20180726] *
JOURNAL OF CLINICAL ONCOLOGY, vol. 34, no. 15, May 2016 (2016-05-01), pages 2582 - 2582
M. MARTINEZ-GARCIA ET AL: "First-in-Human, Phase I Dose-Escalation Study of the Safety, Pharmacokinetics, and Pharmacodynamics of RO5126766, a First-in-Class Dual MEK/RAF Inhibitor in Patients with Solid Tumors", CLINICAL CANCER RESEARCH, vol. 18, no. 17, 3 July 2012 (2012-07-03), US, pages 4806 - 4819, XP055497304, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-12-0742 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11517573B2 (en) 2019-09-13 2022-12-06 The Institute Of Cancer Research: Royal Cancer Hospital Therapeutic compositions, combinations, and methods of use
US11873296B2 (en) 2022-06-07 2024-01-16 Verastem, Inc. Solid forms of a dual RAF/MEK inhibitor

Also Published As

Publication number Publication date
DK3709996T3 (da) 2024-03-11
WO2019096449A1 (en) 2019-05-23
EP3709996A1 (en) 2020-09-23
SG11202004376TA (en) 2020-06-29
US11400090B2 (en) 2022-08-02
EP4364807A2 (en) 2024-05-08
KR20200096538A (ko) 2020-08-12
AU2018369325A1 (en) 2020-06-18
PT3709996T (pt) 2024-04-15
FI3709996T3 (fi) 2024-03-22
MX2020005006A (es) 2020-10-12
US20230012116A1 (en) 2023-01-12
CN111542317A (zh) 2020-08-14
BR112020009452A2 (pt) 2020-12-22
US20210330670A1 (en) 2021-10-28
TW201922254A (zh) 2019-06-16
EP3709996B1 (en) 2024-02-28
TWI795405B (zh) 2023-03-11
RU2760006C1 (ru) 2021-11-22
CA3082619A1 (en) 2019-05-23

Similar Documents

Publication Publication Date Title
US20230012116A1 (en) Coumarin derivative for therapy or prophylaxis of a cell proliferative disorder
US11266654B2 (en) Apilimod compositions and methods for using same
US20190060275A1 (en) Administration of hypoxia activated prodrugs and antiangiogenic agents for the treatment of cancer
US9814724B2 (en) Antitumor effect potentiator and antitumor agent
JP2014509657A5 (zh)
US10350213B2 (en) Methods for treating cancer using apilimod
JP2016508991A5 (zh)
US20150005262A1 (en) Hypoxia activated prodrugs and mtor inhibitors for treating cancer
US20190209576A1 (en) Apilimod for use in the treatment of colorectal cancer
WO2012135779A1 (en) Combinations of akt and mek inhibitor compounds, and methods of use
JP2024056686A (ja) クマリン誘導体を含有する、細胞増殖性疾患の治療又は予防用医薬
JP2016520665A (ja) Mps−1キナーゼ阻害剤および有糸分裂阻害剤を含む、癌の治療のための組合わせ
US20170172985A1 (en) Veliparib in combination with carboplatin and paclitaxel for the treatment of non-small cell lung cancer in smokers
US20210145834A1 (en) Combination of poziotinib with cytotoxic agent and/or other molecularly targeted agent and use thereof
KR102669822B1 (ko) 세포 증식성 질환의 치료 또는 예방을 위한 쿠마린 유도체
CN115038447A (zh) 用于治疗癌症的组合疗法
WO2023011415A1 (zh) Egfr抑制剂的药物组合及其应用
JP6116482B2 (ja) ガリウム錯体を用いる併用療法

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2018560956

Country of ref document: JP

Kind code of ref document: A

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17798235

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: JP

122 Ep: pct application non-entry in european phase

Ref document number: 17798235

Country of ref document: EP

Kind code of ref document: A1